You are on page 1of 12

TYPE Review

PUBLISHED  10 July 2023


DOI 10.3389/fnut.2023.1209238

A narrative review on the use of


OPEN ACCESS probiotics in several diseases.
Evidence and perspectives
EDITED BY
Deep Jyoti Bhuyan,
Western Sydney University, Australia

REVIEWED BY
Mahsa Jalili, Daniela Campaniello †, Antonio Bevilacqua †, Barbara Speranza ,
University of Copenhagen, Denmark Angela Racioppo , Milena Sinigaglia  and Maria Rosaria Corbo *
Naser Alsharairi,
Griffith University, Australia Department of Agriculture, Food, Natural Resources and Engineering, University of Foggia, Foggia, Italy
*CORRESPONDENCE
Maria Rosaria Corbo
mariarosaria.corbo@unifg.it Gut microbiota is a complex ecosystem, strictly linked to health and disease, as a
These authors have contributed equally to this
† balanced composition (referred as eubiosis) is necessary for several physiological
work functions, while an unbalanced composition (dysbiosis) is often associated to
RECEIVED 21April 2023 pathological conditions and/or diseases. An altered microbiota could be positively
ACCEPTED 26 June 2023 affected and partially restored through probiotic supplementation, among others.
PUBLISHED 10 July 2023
This review addresses the effects of probiotics in several conditions, used as case-
CITATION studies (colorectal cancer, neuro-psychiatric diseases, intestinal diseases, obesity,
Campaniello D, Bevilacqua A, Speranza B,
Racioppo A, Sinigaglia M and Corbo MR (2023)
diabetes, metabolic syndrome, immune system, and musculoskeletal system
A narrative review on the use of probiotics in disorders) by pointing out the clinical outcomes, the mode of action, mainly
several diseases. Evidence and perspectives. related to the production of short chain fatty acids (SCFA), the impact of probiotic
Front. Nutr. 10:1209238.
doi: 10.3389/fnut.2023.1209238
dose and mode of supplementation, as well as trying to highlight a hit of the most
used genera.
COPYRIGHT
© 2023 Campaniello, Bevilacqua, Speranza,
Racioppo, Sinigaglia and Corbo. This is an KEYWORDS
open-access article distributed under the terms
of the Creative Commons Attribution License probiotics, disease, clinical trials, effects, genera
(CC BY). The use, distribution or reproduction
in other forums is permitted, provided the
original author(s) and the copyright owner(s)
are credited and that the original publication in
this journal is cited, in accordance with
accepted academic practice. No use,
distribution or reproduction is permitted which
does not comply with these terms.
1. Introduction
Since 2001, Lederbergh and McCray highlighted the importance of microorganisms
inhabiting the human body in health and disease; in fact, a close connection between the “state
of health” of microbial communities and human health was recognized as a milestone (1, 2).
Nowadays, “the assemblage of microorganisms (bacteria, archaea, eukaryotes, and viruses)
present in a defined environment” is called Microbiota (3) and its composition changes
according to the surrounding environment. In particular, the microbiota of the gastro-intestinal
tract, generally known as gut microbiota, is a complex ecosystem composed of fungi, viruses,
and bacteria, adapted to live on the mucus surface of the intestine or in its lumen, affected,
among others, by the modality of childbirth (vaginal vs. cesarean), initial nutrition (breastfeeding
vs. formula) and by the guest genotype (4).
The microbial ecosystem balance is called eubiosis and this status allows to perform several
functions (nutritional, immunological, preventive actions, etc.); but, if this balance is lacking or
altered, there is a condition of “dysbiosis.” Dysbiosis status is often associated to various diseases,
such as asthma, chronic intestinal diseases, obesity, diabetes mellitus, psychiatric disorders, and
many others (5). Several factors, such as antibiotics, smoking, alcohol, a sedentary life, diets low
in fiber, poor chewing, psychophysical stress, chemotherapy, or abuse of drugs (laxatives,
antidepressants, sleeping pills, analgesics) heavily affect microbiota balance and could lead to a
dysbiotic status (6). An altered microbiota could be positively affected and partially restored
through correct diet, and physical activity, although sometimes a supplementation of probiotics
and/or prebiotics (e.g., fibers) could be necessary (7).

Frontiers in Nutrition 01 frontiersin.org


Campaniello et al. 10.3389/fnut.2023.1209238

According to the definition of Food and Agriculture Organization/ the level of signaling pathways, thus they cause an increase of the
World Health Organization (8), slightly modified by Hill et al. (9), mucus, an enhanced production of defensins and proteins in the tight
probiotics are “Live microorganisms which when administered in junctions (11). Finally, probiotics, could act on the immune systems,
adequate amounts confer a health benefit on the host.” They represent through its direct modulation or indirectly acting on gut microbiota.
a strategy to treat intestinal dysbiosis, as they could exert some It has been reported that 70% of immune cells are in the intestine,
important functions, that is (i) anti-inflammatory activity, essential for mainly in the small bowel, where they constitute the gut associated
maintaining the immune response; (ii) to prevent the colonization by lymphoid tissue (GALT) (11), thus suggesting that gut is the main site
pathogenic microorganisms thanks to the physical barrier function; of interaction between host immune systems and commensal
(iii) to produce antimicrobial substances (10). Thousands of authors microorganisms, either positive or pathogenic. Generally, the
studied probiotics and their effects on a wide variety of conditions; a activation of the immune system is first based on the recognition of
search done on Scopus using two keywords (probiotics and disease) PRRs (pattern recognition receptors) by the microbial associated
revealed for 2022–2023 more than 4,000 papers (research papers or molecular patterns (MAMPs); MAMPs are components of microbial
reviews). The analysis of keywords and abstracts through VosViewer, surface able to interact with the gut epithelium and stimulate the cells
a tool for networking and clustering of citations and reference details, of the gut immune system at the lamina propria level (11). Therefore,
pointed out a cluster linked to the effects of probiotics on many T lymphocytes are activated, and helper T lymphocytes (Th) are
diseases (red clusters in Figure 1), including among others diabetes, differentiated, by favoring pro- or anti-inflammatory cytokines
liver diseases, cancer, neurological diseases, obesity etc., thus production (11).
suggesting the interest toward this topic, also stressed by an overview Generally, an eubiotic gut microbiota and probiotics positively
on clinicaltrials.gov. When the search on this database was done (April affect both host’s innate and adaptive immunity (12); concerning
2023), there were more than 2000 items, addressing more than 900 innate immunity, gut microbiota acts both locally and systemically, by
conditions, mainly in Europe and United States (Figure 2). influencing the development and function of antigen presenting cells
The papers available on PubMed, and Scopus have some common (APCs), neutrophils and other innate cell types (12). Moreover, it has
keywords (intestinal flora, gut microbiota, microbiome) and generally been reported the ability of gut microbiota and of some probiotics to
postulate that the beneficial effect of probiotics relies upon the affect innate immunity outside the gut milieu, for example by
modulation of gut microbiota. In addition, another mode of action of promoting the attenuation of inflammation processes at local levels
probiotic into the gut is connected to the improvement of gut barrier (13, 14). There is also a role on adaptive immunity, due to the effect in
mucosa; in fact, both an eubiotic gut microbiota and probiotics act at the development of the most important subtypes of CD4+ T cells (or

FIGURE 1
Clustering and most frequent keywords for the research papers and reviews published in 2022 and 2023 on the effects of probiotics on several disease.
Elaboration through the software VosViewer.

Frontiers in Nutrition 02 frontiersin.org


Campaniello et al. 10.3389/fnut.2023.1209238

FIGURE 2
Studies on probiotics on clinicaltrials.gov.

helper T cells, which are lymphocytes coordinating the response to cell differentiation (22). In addition, tryptophan (deriving from diet)
diseases), that is Th1, Th2, Th17 and Treg (12, 15). In addition to T cells, and indolic acid derivatives (for example IPA, indole-3-propionic
an eubiotic gut microbiota could influence B cell maturation and acid) bind to receptors expressed on immune cells, promote IL-10
immunoglobulin production (16). production with anti-inflammatory activity and decrease TNF-α
The mechanisms by which gut microbiota and probiotics release (21).
influence immune system include the production of various It is worth mentioning that the ability of potential probiotics to
compounds; SCFA (short chain fatty acids; butyrate, acetate, formate), modulate the immune system and ameliorate inflammatory status
indole derivatives, and bile salts are, among others, the most depend on the strains and a comprehensive overview of the effects at
important. An extensive description of the effects of indole derivatives species level is missing (23). Other topics missing in the literature are
on gut microbiota is in the review of Ye et al. (17); however, it is worth the technological aspects of the problems (production and dose of
mentioning that indole derivatives, produced by gut microbes and probiotics). Therefore, the main goal of this paper is an overview of
some probiotic strains (e.g., Limosilactobacillus reuteri) through the the effects of probiotics on some representative conditions, addressing
metabolism of tryptophan are crucial, because they enhance intestinal some key-points, like the clinical effects, and the mode of action of
epithelial cell function by regulating several genes involved in probiotics, if available; the elucidation of aspects common to all strains
mechanical barrier formation. Moreover, they increase mucin and of a species, and finally a focus on the importance of a correct dose.
goblet cell secretion products, responsible of barrier of gut mucosa, There are many pathological conditions; however, by authors’
and reduce the impact of possible pathogens (17). choice only research papers and some representative conditions were
SCFA are produced through the fermentation of non-digestible chosen, as best models for future studies, that is colorectal cancer,
carbohydrates and amino acids in the colon and play a major role in neuro-psychiatric diseases, intestinal diseases, obesity, diabetes,
maintaining the barrier function of gut (18). They are absorbed by the metabolic syndrome, which are probably the most addressed topics in
colonocytes and used as fuel for the colonic mucosal epithelial cells the literature, along with two minor issues (immune system, and
(19), but at the same time they directly act on gut mucosa; for example, musculoskeletal system disorders), which are promising ways but with
butyrate contributes to reduce oxidative stress, thus stabilizing gut a few evidence.
mucosa and reducing the translocation of LPS (Lipopolysaccharide) For each pathological conditions, the effects of probiotics are
(12). Also, bile salts are essential for immunity in a bidirectional described, and the list of studies and outcomes is in reported, along
crosstalk between host and microbiota. Primary bile salts, or host- with the kind of probiotic, or the probiotics mix, the target of the study
derived bile salts, shape and modify the composition of microbiota, (humans or animal model), and the achievable and measurable
generally reducing the levels of Gram-negative bacteria; while those outcomes (Supplementary Table S1).
synthesized by microbiota contribute to a further modulation of
microbiota itself and act on both innate and adaptive immunity, for
example by reducing the levels of pro-inflammatory cytokines, or 2. Colorectal cancer
enhancing Treg cells differentiation (20).
SCFA and derivatives from tryptophan could also play a Colorectal cancer (CRC) is the most frequent neoplastic form of
significant role in reducing inflammatory status. SCFA bind to specific the gastrointestinal tract; its incidence is experiencing a progressive
receptors on intestinal epithelial cells, thus they inhibit NF-κB increase, due to a gradual aging of the population, the adoption of
pathway, Treg cell suppression, and pro-inflammatory cytokine sedentary lifestyle, and unbalanced diets (24), as also suggested by the
production by neutrophils and macrophages (21). For example, higher incidence rates in Australia and New Zealand, North America,
butyrate could control gut inflammation through the induction of Treg and Europe (25). Although it is a multi-etiological condition, it should

Frontiers in Nutrition 03 frontiersin.org


Campaniello et al. 10.3389/fnut.2023.1209238

be considered the genetic susceptibility of each individual, as well as similar effects could be observed for Lactobacillus acidophilus MTCC
some environmental factors connected to carcinogenesis, like caloric 5401 (42) and Faecalibacterium prausnitzii (43), while Lactococcus
intake, obesity, alcohol or smoking (26–37). Focusing on gut lactis subsp. cremoris C60 and Lacticaseibacillus casei ATCC334
microbiota, CRC patients often develop a dysbiosis due to the use of probably exerted a preventive and an inhibitory effect on the cells
antibiotics, radiation therapy, and chemotherapy, and their gut responsible for CRC (44, 45). In addition, an emergent butyrate-
microbiota is characterized by an increased pathogenic bacteria producing probiotic, Butyricicoccus pullicaecorum exerted antitumor
abundance, decreased SCFA-producing bacteria and SCFA levels (38, effect and showed good acid and bile tolerance; it was also able to
39) and butyrate seems the most affected compound, as it could reduce pathogen population and to prevent necrotic enteritis (41).
be successfully used as a potential biomarker of CRC risk or as an early Shang et al. (46) demonstrated the effectiveness of a probiotic mix
warning signal of the disease onset (40). Conversely, high levels of composed of Bifidobacterium longum, Bifidobacterium bifidum,
SCFA have antineoplastic properties, due to a combination of several L. acidophilus and Lactiplantibacillus plantarum in mice, able to
mechanisms, like the downregulation of the canonical Wnt signaling reduce the tendency of CRC cells to migrate in different body tissues.
pathway linked to colonic carcinogenesis, the limitation of Furthermore, the tumor size in mice feed with probiotic mixture was
proliferation and migration of neoplastic cells, the suppression of significantly smaller than the control group. In another study, Dong
tumor angiogenesis, the induction of apoptosis and the promotion of et al. (47) investigated Ligilactobacillus salivarius effect on CRC cells,
neoplastic colonocytes differentiation (40). via oral administration in male mice. The authors reported that
Although the production of SCFA probably exerts a major role in the probiotic induced the suppression of dimethylhydrazine (DMH)
anti-carcinogenic activity of probiotics, there are also some other direct production, both in the early and post-early stages of carcinogenesis.
and indirect effects, briefly summarized in Figure 3, including the ability DMH is a potent carcinogen used to induce colon cancers in animals,
to catch and adsorb carcinogenic compounds, as well as by stimulating particularly mice. These results therefore suggest that daily oral
host’s antitumor activity through the stabilization of the tight junctions administration of L. salivarius could effectively prevent CRC
or the production of defensins. Other effects include the antagonistic carcinogenesis by inhibiting cell proliferation and inducing apoptosis
activity toward putrefactive microbiota and the creation of a in DMH-induced tumor models.
microenvironment into the colon unfavorable for the carcinogenesis. Probiotics could also counteract dysbiosis occurring in most
Many research papers and clinical trials have addressed the role patients after CRC resection and improve the biodiversity of bacterial
of probiotics in the CRC onset and/or mitigation and a comprehensive biota. In this context, Park et  al. (48) observed improvements in
overview of the most important trials is in the paper of Hou et al. (40); postoperative intestinal dysbiosis with the use of probiotics in CRC
Supplementary Table S1 shows some relevant studies. In particular, surgical resection patients. Sixty patients, aged between 18 and 75,
Bacteroides fragilis exerts anti-inflammatory and anticancer effect, as with sigmoid colon adenocarcinoma and anterior resection of the
it can alter the composition of the microbiota, inactivating same, were divided into two groups: 29 and 31 patients feed with a
carcinogenic compounds, competing with pathogens or probiotic mixture (Bifidobacterium animalis subsp. lactis HY8002,
CRC-promoting bacteria and stimulating the immune response (41); L. casei HY2782 and L. plantarum HY7712) and the placebo

FIGURE 3
Probiotic effects on CRC.

Frontiers in Nutrition 04 frontiersin.org


Campaniello et al. 10.3389/fnut.2023.1209238

respectively, for 4 weeks. Probiotics led to an increased production of Streptococcus genera as well as L. plantarum PS128, Limosilactobacillus
SCFA by colon bacteria, decreased microbes associated with the reuteri and Lacticaseibacillus rhamnosus GG (56–62). In particular, the
development of CRC (mainly Alloprevotella and Porphyromonas) and effectiveness of L. plantarum PS128 relied upon the age of the children,
improved postoperative recovery of patients. Particularly interesting as the best results were obtained on infants (60).
were the data obtained from the measurement of faecal zonulin, a For anxiety and depression, the outputs showed an improvement
protein that acts on the tight junctions of the intestine, regulating its in the gut microbiota with a reduction in depressive and anxious
permeability; high levels are associated with a deterioration of the behavior (63–65). In particular, Abildgaard et  al. (64) proposed a
intestinal mucosa, which does not adequately perform its protective mixture of probiotics (B. bifidum W23, Bifidobacterium lactis W52,
function. The authors found that zonulin significantly decreased in the L. acidophilus W37, Levilactobacillus brevis W63, L. casei W56,
group fed with probiotic mixture compared to the placebo group. L. salivarius W24, Lactococcus lactis W19, L. lactis W58) as potential
The efficiency of L. plantarum was also observed by Yoon et al. treatment strategy in major depressive disorders (MDD) to reduce
(49); the authors evaluated the effect of L. plantarum CJLP243 depressive behavior. Some studies reported improvement in behavioral
(isolated from kimchi, a traditional fermented product of Korea) on abnormalities and reduction in the main symptoms of depression in
intestinal function and quality of life toward 36 patients aged 20–75, humans, after the administration of strains belonging to the genera
who have undergone rectal resection and were admitted undergoing Lactobacillus and Bifidobacterium (66–71). Another possible use of
the reversal of the ileostomy. Unfortunately, a significant number of probiotics refers to dementia and cognitive deterioration. The intake
patients reported symptoms including diarrhea, fecal incontinence, of Enterococcus faecium together with inulin (72) and Bifidobacterium
and other complications. The patients were divided into two groups: breve A1 (73) improved learning and memory skills, language,
19 and 17 patients who took placebo and probiotic respectively, once attention and orientation in the elderly people. In addition, some
a day for the duration of 3 weeks. The results showed that there were studies on animals showed an improvement in the intestinal barrier
no significant differences between the two groups regarding the and spatial learning through the administration of L. casei LC122, of
improvement of symptoms; however, by comparing the post-operative B. longum BL986 and of Clostridium butyricum (74, 75).
results between the first and third weeks, the administration of the For Parkinson’s disease (PD) Tamtajii et al. (76) and Magistrelli
probiotic showed a tendency to improve intestinal function and et  al. (77) observed that L. acidophilus, B. bifidum, L. reuteri,
quality of life. Limosilactobacillus fermentum and L. salivarius allowed an
improvement in MDS-UPDRS (Movement Disorder Society-Unified
Parkinson’s Disease Rating Scale) scores and a significant reduction in
3. Neuro-psychiatric diseases pro-inflammatory cytokine levels and reactive oxygen species (ROS),
with a possible weight of the stage of the disease and sex. In animal
Many human and animal studies support the idea that gut models, Barichella et al. (78) showed that the genera Lactobacillus and
microbiota plays an important role for cognitive functions, in the Bifidobacterium could improve intestinal integrity and reduce anxiety,
regulation of mood and emotions, and in the interpersonal depression and stress.
interactions and communications (50). Gut microbiota can modulate Anorexia nervosa (AN) consists of an altered perception of one’s
brain activity and behavior; therefore, its manipulation can be applied own body, in particular weight. In fact, people who are in this
in the treatment of neuropsychiatric disorders such as autism condition try to keep their body weight as low as possible through a
spectrum disorders, depression, etc. (51, 52). The idea that probiotic strong dietary restriction, inducing vomiting and practicing intense
could positively affect the clinical outcomes of depression was first physical activity. AN most frequently affects young women, although
postulated in 1910 when Hubert J. Norman and Georges Porter recently it has also targeted men; it can often be  associated with
Philipps found an improvement in the symptoms after taking psychological problems such as depression, anxiety, low self-esteem,
lactobacilli (53). Later then, this idea has been confirmed by several alcohol abuse, and self-harm (79, 80). L. plantarum P8 determined a
studies and clinical trials, although the mode of action of probiotic on reduction in anxiety and stress (81) while B. fragilis reduced gastro-
behavior and neuro-psychiatric diseases is still unclear, as in some intestinal pains and caused as a secondary effect an increase serotonin
cases symptoms improvement and amelioration are not related to a production (82); it is not clear if these effects have a connection or are
modification in gut microbiota (53). independent outcomes (Supplementary Table S1). In animals,
Supplementary Table S1 reports 33 scientific articles concerning Lactobacillus spp. promoted weight gain (83) and improved the
the effect of probiotics in subjects with neuro-psychiatric diseases. behavioral abnormalities in stressed mice involving the microbiota-
Twelve articles refer to autism (ASD), a neurobiological developmental brain gut axis (84). Moreover, Akkermansia muciniphila, considered a
disorder, characterized by severe and generalized impairment of both potential candidate for improving metabolic disorders associated with
communication skills and social interaction. Subjects affected by ASD, anorexia, obesity, diabetes, liver disease, favored the restoration of a
especially in children aged 2 to 11 years, show a stereotypical use of compromised intestinal barrier (85).
movements, language or objects, excessive adherence to routine Probiotics were also studied in relation to the benefits they bring for
situations, routines, rituals, and fixation for particular or restricted other diseases affecting the brain systems. For example, the
interests abnormally in duration or intensity (54). The benefits of administration of L. acidophilus, B. bifidum and B. longum, improved the
probiotics depend on the microorganisms. For example, an anti- cognitive function of Alzheimer’s patients (humans and in animals) (86,
inflammatory effect was found following the administration of 87), while strains of L. rhamnosus GG and B. animalis subsp. lactis Bb12
Bifidobacterium spp. (55), while improvement of gastrointestinal led to an improvement of the symptoms related to schizophrenia (such
disorders and neuro-behavioral symptoms was achieved by microbial as delirium, hallucinations, language, and disorganized behavior, etc.)
mixtures composed of several strains of Bifidobacterium, Lactobacillus, (88). Furthermore, in women aged 20–40 affected by multiple sclerosis,

Frontiers in Nutrition 05 frontiersin.org


Campaniello et al. 10.3389/fnut.2023.1209238

a mixture of probiotics (Lacticaseibacillus paracasei, L. plantarum, role in the pathophysiology of obesity. Firmicutes and Bacteroidetes
L. acidophilus, L. delbrueckii, B. longum, Bifidobacterium infantis, are the two phyla involved in microbial dysbiosis and in the
B. breve, Streptococcus thermophilus) improved the symptoms by development of obesity. The ratio between these phyla is very
modulating the anti-inflammatory immune response (89). Referring to important; in fact, Bervoets et al. (102) studied the gut microbiota of
multiple sclerosis, Altieri et al. (90) in a recent review described how 26 overweight and obese children and 27 skinny children and found
microbiota change in MS patients and proposed probiotics as useful that obese children have a higher ratio of Firmicutes to Bacteroidetes.
tools to improve the symptoms of MS patients. Supplementary Table S1 focuses on some application of probiotics
toward overweight and obese subjects. Kadooka et  al. (103)
administered fermented milk containing Lactobacillus gasseri
4. Intestinal diseases SBT2055 (200 g/day) to 87 overweight adults for 12 weeks. Reductions
in visceral and subcutaneous fat, body weight and BMI (Body Mass
Generally, probiotics could positively impact on gastrointestinal Index) compared to the control group, were observed. Furthermore,
disorders (GI) (abdominal pain or discomfort, swelling and flatulence) the consumption of yogurts supplemented with capsules, containing
through metabolic effects resulting from enzymatic activity and the 109 CFU of Lactobacillus amylovorus and L. fermentum by 28
crosstalk with the central nervous system, by improving gut function overweight participants, led to a reduction in total body fat mass
(91). In addition, there are several evidence on positive effects on (104). Regarding gut microbiota, the researchers observed a significant
Inflammatory Bowel Disease (IBD) and Irritable Bowel reduction of Clostridium cluster IV (for L. amylovorus consumption),
Syndrome (IBS). together with an increase of Lactobacillus in both treatments and
Concerning IBD, Ferreira-Halder et al. (43) and Lopetuso et al. concluded that when the gut microbial composition is modulated
(92) highlighted the anti-inflammatory effect performed by through probiotic consumption, this can positively alter energy
F. prausnitzii and A. muciniphila. F. prausnitzii contributes metabolism and body composition (104). An additional study on 70
substantially to the health of the intestine and is considered a overweight and obese children revealed that a combination of
biomarker not only for human health but also for diagnosis and probiotics, prebiotics and vitamins A, E and C for 8 weeks, significantly
subsequent treatment (43). On the other hand, A. muciniphila has reduced BMI, waist circumference, waist/hip ratio, LDL cholesterol
been shown to be effective in immune and metabolic regulation; it and triglycerides (105).
ensures increased function of the intestinal barrier showing a direct Probiotics can reduce cholesterol levels through bile salt hydrolase
and beneficial effect on the host’s response. In addition, its use is (an enzyme that hydrolyzes bile salts into amino acid residues and free
considered safe if aimed at human studies (93). bile salts). 200 g/day of yogurt containing S. thermophilus, L. delbrueckii
In patients with ulcerative colitis, probiotics act as a barrier against subsp. bulgaricus, L. acidophilus LA-5, and B. animalis BB12 for
harmful microorganisms. A consortium of 8 probiotic strains (VSL3, 9 weeks to 70 women in the third trimester of pregnancy resulted in
composed of L. casei, L. plantarum, L. acidophilus, L. delbrueckii subsp. significant reductions in total cholesterol, LDL cholesterol, and high-
bulgaricus, B. longum subsp. longum, B. breve and B. longum subsp. density lipoprotein (HDL) cholesterol, as well as serum triglyceride
infantis, Streptococcus salivarius subsp. thermophilus) was effective in concentrations (106). Probiotic supplementation also reduced blood
maintaining a state of remission (94), while Azad et al. (95) reported lipid concentrations (107).
that Lb. acidophilus restored the balance of inflammatory cytokines A. muciniphila administered to animals led to a reduction in fat
and Th17/Treg cells in mice induced colitis, and showed beneficial mass and body weight; moreover, it favored the restoration of the
effects in the prevention of cancer and intestinal inflammation (95). intestinal barrier function and, if administered to humans,
In addition, several analyses have shown the effectiveness of the improved inflammation, insulin resistance and blood sugar
administration of probiotics in premature infants, with a reduction of level (108).
both the development of enterocolitis and the risk of sepsis in old age. Many authors reported that the action of probiotic toward obesity
In particular, Dermyshi et  al. (96) supported the benefits of is mediated by SCFA, which probably could be  involved in body
L. acidophilus-B. infantis blend. weight regulation, and maintenance, as well as in energy intake and
IBS causes swelling, vomiting, diarrhea, abdominal pain, expenditure (109–111). Although there are several hypotheses, the
frequency of stools, and probiotics could improve these symptoms. most probable mechanism involves the ability of propionate and
Two formulations containing different probiotic strains butyrate to bind to G-protein-coupled receptors in the colon leading
(F1 = L. acidophilus, L. reuteri; F2 = L. plantarum, L. rhamnosus, to the production of the gut hormones peptide YY and glucagon-like
B. animalis subsp. lactis) were administered to humans, thus gaining peptide 1, thus influencing satiety and glucose homeostasis (109). In
a relief in bloating, abdominal pain, constipation, abdominal cramps, addition, SCFA activate intestinal gluconeogenesis, and the released
and flatulence (97). Similar effects were observed through the glucose mediates a signal to brain through portal nerves for satiety
administration of Bacillus coagulans MTCC 5856 (98), and and insulin sensitivity, or they can also affect peripheral metabolism
L. plantarum DSM 9843 (99). Other studies reported the improvement in the liver (enhanced lipid oxidation, lower lipid storage), skeletal
of IBS symptoms due to several lactobacilli (100, 101). muscles (increase of glycogen synthesis and reduction of glycolysis),
pancreas (increase of insulin and reduction of glucagon synthesis and
release) or adipose tissue (reduction of insulin mediated adiposity)
5. Obesity (109, 110). The evidence available in the literature suggest that that
increasing SCFA production could be  a preventive measure to
Gut microbiota is involved in the control of body weight, energy counteract gastro-intestinal dysfunction, obesity, and type 2 diabetes
homeostasis and inflammation states; therefore, it plays an important mellitus (109, 110), although longer term trials and data are required,

Frontiers in Nutrition 06 frontiersin.org


Campaniello et al. 10.3389/fnut.2023.1209238

also to elucidate the exact role of the initial imprinting of gut and improved liver. Finally, CPCM increased LAB and Bifidobacterium
microbiota and how it can respond to probiotic intervention. population in intestinal tract and decreased Escherichia.
Razmpoosh et  al. (118) evaluated the effect of 7 probiotics
(L. acidophilus, L. casei, L. rhamnosus, L. bulgaricus, B. breve,
6. Diabetes B. longum, S. thermophilus), and 100 mg of fructo-oligosaccharide
(FOS) with lactose as carriers, on lipid profile and glycemic control in
Generally systemic inflammation involve microbiota as it 60 patients. They were equally divided into 2 groups (group 1 took
modulates inflammation, interacts with nutrients, influences intestinal probiotics and group  2 took a placebo, for 6 weeks). A significant
permeability, glucose and lipid metabolism, insulin sensitivity and the decrease in the fasting plasma glucose (FPG) and increase of high
body’s energy balance. The microbiota of diabetic patients is poorly density of lipoprotein cholesterol (HDL-C), was observed. No
populated by useful microorganisms (Bifidobacterium, Bacteroides, significant differences in the levels of insulin, triglycerides, total
Faecalibacterium, Akkermansia, and Roseburia) which have anti- cholesterol, insulin resistance and anthropometric measurements
inflammatory activity, are butyrate-producing and are promoters of (weight, waist circumference and body mass index).
low intestinal permeability and may have inhibitory activity against
carbohydrates-degrading enzymes, reducing postprandial
hyperglycemia. On the contrary, there are many microorganisms 7. Metabolic syndrome
favoring the production of inflammatory molecules and the alteration
of intestinal permeability such as Ruminococcus, Fusobacterium, and Metabolic syndrome (MetS) is a pathology characterized by an
Blautia (112, 113). In any case, considering that diabetes is closely excess in abdominal fat, arterial hypertension, impaired fasting plasma
linked to food choices and habits, it is certainly essential to make glucose (FPG) or insulin resistance, whose diagnoses and treatments
adequate decisions in this regard; for example, an active lifestyle could are often similar to those of obesity (119). Supplementary Table S1 lists
improve insulin resistance, while taking foods rich in fibers, largely 6 papers concerning the study of the effect of some probiotics in
represented by prebiotics, is certainly a positive choice for subjects with MetS.
wise prevention. Corb Aron et al. (108) and Ottman et al. (93) used A. muciniphila
Positive effects such as increased insulin sensitivity and to evaluate its effect on volunteers with MetS. They observed that the
improvement of microbial diversity were found following probiotic degrades mucin by stimulating the production of new
administration of L. reuteri DSM 17938 to patients with type 2 mucous layer (108) and contributes to immune and metabolic
diabetes (114). regulation by increasing the intestinal barrier (93). At the same time,
Toejing et al. (115) administered L. paracasei HII01 (50 × 109 CFU/ the metabolic activity of A. muciniphila led to the production of SCFA
day) to 50 T2DM (type 2 diabetes mellitus) patients to evaluate the with beneficial effect to the host and members of the microbiota (93).
effect on glycemia and observed that after 12 weeks fasting blood Instead L. plantarum (120), L. acidophilus and some
glucose (FBG) level significantly decreased. Furthermore, probiotics Bifidobacterium species (B. bifidum, B. lactis, and B. longum) (121)
reduced the plasma levels of lipopolysaccharide (LPS), inflammatory mainly led to a reduction in blood sugar and cholesterol. In particular,
markers (TNF-α, IL-6) and C-reactive protein (hsCRP). A reduction reduction in LDL cholesterol, blood glucose, and homocysteine ​​levels
in pathogenic microorganisms together with improvement in when postmenopausal women were treated with L. plantarum for
beneficial bacteria were also observed; therefore, the authors 90 days (120).
concluded that L. paracasei HII01 could play a potential role as an
adjuvant treatment in type 2 diabetes.
A potential antidiabetic effect was also observed by using another 8. Musculoskeletal system
Lactobacillus strain: Wu et al. (116) investigated the performances of
L. rhamnosus LRa05 on glucose metabolism and gut microbiota in The role of probiotics in the control of musculoskeletal diseases is
T2DM mice. The treatment with 109 CFU/day of L. rhamnosus resulted a topic of great interest; osteoporosis (characterized by a decrease in
in a reduction in the fasting blood glucose (FBG) levels (by 53.5%), bone strength, a low mineral density of the bone tissue, with
lowered insulin resistance, alleviated metabolic lipopolysaccharide- consequent fragility and aging) (122), osteoarthritis (a
related inflammation and relieved hepatic oxidative stress. Further non-inflammatory arthropathy involving cartilage and bone
positive effects were found on the gut microbiota composition; in fact, remodeling) or bone fragility, and microbiota changes are closely
SCFA producing microorganisms, such as Alloprevotella and related (123).
Bacteroides, increased with a reduction of proinflammatory It has been demonstrated that the synergistic action of L. casei
microorganisms such as Odoribacter and Mucispirillum (116). with type II collagen (CII) and glucosamine (GS) (potential prebiotic),
Manaer et  al. (117) reported the benefits of Lactobacillus and administrated to arthritic rats, led to an effective reduction of pain and
yeasts on T2DM mice. Probiotics (Lactobacillus kefiranofaciens, cartilage destruction. Moreover, a reduced expression of numerous
L. plantarum, Lactobacillus helveticus, L. lactis, Issatchenkia orientalis), proinflammatory cytokines, resulted (124).
isolated from traditional fermented cheese whey (TFCW), were used Supplementary Table S1 reports some cases concerning the use of
to prepare a mix from camel milk (CPCM) to feed db/db mice. The different Lactobacillus strains to relieve bone, joint and muscle
authors studied how these strains affect gut microbiota, glucose and disorders. The ability of probiotics to reduce pain and cartilage
lipid metabolism, liver and renal functions. CPCM reduced fasting destruction has been highlighted in experiments conducted on
blood glucose (FBG), oral glucose tolerance test and glycosylated animals (125) together with numerous effects, such as antimicrobial,
hemoglobin HbAlc, increased C-Protein, modulated lipid metabolism antioxidant, anti-inflammatory (126), the ability to determine an

Frontiers in Nutrition 07 frontiersin.org


Campaniello et al. 10.3389/fnut.2023.1209238

increase in calcium (127) and recovery of joint strength (128)


in humans.
Steves et al. (125) and Paul et al. (126) demonstrated that L. casei
and L. acidophilus improved intestinal dysbiosis and the symptoms of
rheumatoid arthritis after long-term repeated use thanks to their anti-
inflammatory, antimicrobial and antioxidant properties. These
microorganisms act symbiotically in the intestine to establish their
colonization and consequently increase the integrity of the cell layers
of the gastro-intestinal tract, maintain the nutritional support of the
host and reduce the severity of inflammatory conditions.

9. Immune system disorders


It is known that probiotics can also bring benefits through the FIGURE 4
modulation of the immune system. Supplementary Table S1 shows 3 Probiotic genera mostly used in clinical trials.
articles focused on the effect of probiotics on the modulation of the
immune system. Among the most significant results, there are the
bactericidal and antitumor effect with production of proinflammatory
and anti-inflammatory cytokines in humans, by E. faecium (95) and advises manufacturers to list expected probiotic concentration on the
the development of regulatory cells in the gastrointestinal epithelium “expiration” or “use by” date on the product label when stored at
in animals, by strains of L. reuteri (99). Finally, Han et al. (129) treated proper conditions and suggests consumers to avoid preparations
mice with L. rhamnosus HDB1258 and observed that it enhanced the listing the dose of probiotic at the time of production (132).
immune response by activating innate immunity. In addition, Strictly linked to the dose, the second critical point is the duration
L. rhamnosus suppressed systemic inflammation by increasing the of supplementation, but for this aspect there is not a consensus in the
expression ratio of anti-inflammatory cytokines and modulated the literature; generally, it is believed that probiotics should be assumed
microbiota composition. for several weeks (at least from 2 to 4 weeks) to gain achievable
outputs (133). However, the supplementation could be either short-
term or long-term, with short-term interventions suggested only for
10. Probiotic species, dose, delivery, acute gastro-intestinal conditions (5–7 days for acute diarrhea in
and production infants and children, from 1 to 4 weeks for antibiotic-associated
diarrhea, a few weeks for constipation) (133, 134), while other
This review shows that there are significant effects of probiotics conditions require long-term supplementation, up to 2–3 months for
on a wide variety of conditions; moreover, a focus at genus/species IBD, 3–6 months for Chron disease, atopic dermatitis, or psychiatric
level on research papers with a robust design beyond and with proven diseases (133–136).
effects (ca. 160) suggests the efficacy of lactobacilli (L. plantarum, Another critical point is the delivery. Probiotics are marketed in
L. casei, L. acidophilus, L. reuteri, among others) and bifidobacteria different forms such as capsules, tablets, films, or hydrogels, and for
(B. longum, B. infantis, B. animalis, B. bifidum or B. breve), with oral delivery the microencapsulation in hydroxypropyl methylcellulose
promising evidence for a new generation of probiotics (mainly phthalate (HPMCP), hydroxypropylmethyl cellulose acetated
A. muciniphila, B. fragilis, and F. prausnitzii; Figure 4). Apart from succinate, and cellulose acetate phthalate (CAP) is used to minimize
species, the identification of the dose required to gain a measurable the exposure of probiotics to gastric acids, reducing their viability loss
output is controversial. Many probiotic supplements contain 1 to in the stomach (137). It is a matter of debate if oral delivery mediated
10 billion CFU per dose, up to 50 billion CFU or more; however, by foods could result in a higher impact of probiotics (138), while
higher CFU counts do not necessarily improve health effects. In fact, other ways of delivery, less used at least for the studies reported in this
depending on the disorder, it may happen that even a lower dose can review, are nasal, transdermal, rectal, and vaginal (137).
be  effective or even better than a higher dose (130). Also, production could affect viability and thus health effects of
Supplementary Table S1 shows the doses, when available, for the probiotics; fermentation is the most common method of producing
different trials; generally, the concentrations for the most important commercial probiotics: in a large fermentation vessel, single-strain
commercial preparations of Lactobacillus spp. and Lactobacillus probiotics are inoculated into a liquid broth that is stirred to prevent
related genera are from 109 to 1010 CFU, while for Bifidobacterium bacterial settlement and with pH kept under control. When the
spp. at 108–1010 CFU, for Pediococcus acidilactici 109 CFU, for production concerns anaerobic species, gasses such as nitrogen,
Streptococcus thermophilus 108 CFU, for yeast strains such as hydrogen, and carbon dioxide, are controlled. Microbial growth is
Saccharomyces boulardii 109 CFU, Bacillus subtilis 109 CFU and controlled by cell density measurements and light/fluorescence
A. muciniphila 108 CFU (131). It is worth mentioning that the dose is microscopes are used to check for unwanted contaminations. Once
also a function of storage conditions, as some preparations should batch fermentation is complete, a filtered and concentrated cells
be stored at room temperature, while others require refrigeration; suspension is either spray-dried or freeze-dried but previously,
therefore, a thermal abuse could heavily affect probiotic survival. The cryoprotectants or lyoprotectants are added to prevent loss of
International Scientific Association for Probiotics and Prebiotics microbial viability (131).

Frontiers in Nutrition 08 frontiersin.org


Campaniello et al. 10.3389/fnut.2023.1209238

To increase the production rate the batch fermentation is Moreover, most papers focus on the medical point of view, while
integrated with crossflow membrane ultra/microfiltration; when the there is a dark side not addressed, that is the technological story
desired cell density is reached, toxic metabolites and/or acids are connected to probiotic productions, the way of supplementation (with
removed through a membrane. Fresh medium is continuously food or as supplements), the shelf life, and the dose at the time of
pumped into the fermenter by varying flow rates to ensure a constant consumptions, among others. Further efforts are required to address
total volume. The cell suspension can then be extracted in batches or both medical issues and technological/microbiological challenges for
continuously (131). an effective use of probiotics as concurrent strategies for many
Another effective method to enhance the production of probiotics pathological conditions; there are promising evidence and data, but
is the immobilization in natural biopolymers such as protein-based we are still at a preliminary level, as an effective and efficient use of
biopolymers, polysaccharides, lipids, and synthetic polymers or probiotics should be based on the clear definition of a “before” (dose,
coating for the protection of probiotics against moistures or gasses storage, way of supplementation, duration etc.) and an “after” (outputs
(oxygen/carbon dioxide) (131). Cells are immobilized in clearly evidenced and defined).
polysaccharide hydrogels, then placed in a fermenter, where the
medium is regularly supplemented, and the cells periodically removed
to ensure proper dilution. This strategy is used to improve overall Author contributions
growth rate and cell viability. The benefits of this approach are the
continuous and controlled delivery of probiotics to the gut, a higher MC, DC, and AB: conceptualization. MS, BS, and AB:
viability, and lower costs, while the some limits are the restricted methodology. AR, DC, and AB: investigation and data. AB and DC:
biocompatibility of some immobilization agents, and the complexity writing–original draft preparation. DC, AB, BS, AR, MS, and MC:
of production processes (131). writing–review and editing. MC: supervision. MS and MC: project
administration and funding acquisition. All authors have read and
agreed to the published version of the manuscript.
11. Conclusions and perspectives
The use of probiotics could be a promising strategy to counteract Conflict of interest
side or secondary effects in several pathological conditions; the
evidence and data hereby reported suggest a benefit in CRC both as a The authors declare that the research was conducted in the
preventive measure to avoid carcinogenesis or during medical absence of any commercial or financial relationships that could
treatments to favor recovery, or in improving cognitive functions, in be construed as a potential conflict of interest.
ameliorating the symptoms of some intestinal diseases (e.g., IBD), or
to counteract obesity, diabetes and other metabolic syndromes. The
effect is generally mediated through the modulation of gut microbiota, Publisher’s note
as well as on the production of significant amounts of SCFA, which
exert in turn several physiological functions, and the final output All claims expressed in this article are solely those of the authors
could be symptoms amelioration or disease remission, although the and do not necessarily represent those of their affiliated organizations,
use of different clinical outcomes is a challenge, as it makes difficult a or those of the publisher, the editors and the reviewers. Any product
comparison of different trials and research papers. that may be evaluated in this article, or claim that may be made by its
At species level, most data are available on Lactobacillaceae and manufacturer, is not guaranteed or endorsed by the publisher.
on Bifidobacterium spp., even if evidence is available for A. muciniphila,
B. fragilis, and F. prausnitzii. However, there are some issues that
should be addressed, related to the duration of the supplementation Supplementary material
(short-term or long-term), dose, as each study suggests a different
dose (ranging from 108 to 1010 CFU). Concerning the way of The Supplementary material for this article can be found online
supplementation, oral delivery is preferred, but there is still a debate at: https://www.frontiersin.org/articles/10.3389/fnut.2023.1209238/
on the usefulness of a supplementation through food. full#supplementary-material

References
1. Lederberg J. McCray AT ‘Ome sweet’ omics. A genealogical treasury of words. 5. De Siena M, Laterza L, Matteo MV, Mignini I, Schepis T, Rizzatti G, et al. Gut and
Scientist. (2001) 15:8. Reproductive Tract Microbiota Adaptation during Pregnancy: New Insights for
Pregnancy-Related Complications and Therapy. Microorganisms. (2021) 9:1–15. doi:

2. Aggarwal N, Kitano S, Puah GRY, Kittelmann S, Hwang IY, Chang
10.3390/microorganisms9030473
MW.  Microbiome and human health: Current understanding, engineering, and
enabling technologies. Chem Rev. (2023) 123:31–72. doi: 10.1021/acs. 6. Marangoni F, Poli A. Microbiota intestinale; salute umana e prebiotici. Milano:
chemrev.2c00431 Pacini ED (2017).
3. Marchesi JR, Adams DH, Fava F, Hermes GDA, Hirscfield GM, Hold G, et al. The 7. Campaniello D, Corbo MR, Sinigaglia M, Speranza B, Racioppo A, Altieri C, et al.
gut microbiota and host health: a new clinical frontier. Gut. (2016) 65:330–9. doi: How diet and physical activity modulate gut microbiota: evidence, and perspectives.
10.1136/gutjnl-2015-309990 Nutrients. (2022) 14:2456. doi: 10.3390/nu14122456
4. Capurso L. Il Microbiota Intestinale. Recenti Prog Med. (2016) 107:257–66. doi: 8. FAO/WHO. Food and Agriculture Organization of the United Nations/World Health
10.1701/2296.24680 Organization. Joint FAO/WHO expert consultation on evaluation of health and nutritional

Frontiers in Nutrition 09 frontiersin.org


Campaniello et al. 10.3389/fnut.2023.1209238

properties of probiotics in food including powder milk with live lactic acid bacteria. 33. Leenders M, Siersema PD, Overvad K, Tjønneland A, Olsen A, Boutron-Ruault
Geneva: World Health Organization (2001). MC, et al. Subtypes of fruit and vegetables, variety in consumption and risk of colon and
rectal cancer in the European Prospective Investigation into Cancer and Nutrition. Int
9. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, et al. The International
J Cancer. (2015) 137:2705–14. doi: 10.1002/ijc.29640
Scientific Association for Probiotics and Prebiotics consensus statement on the scope
and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol. (2014) 34. Zhang SM, Moore SC, Lin J, Cook NR, Manson J, Lee I-M, et al. Folate, vitamin
11:506–14. doi: 10.1038/nrgastro.2014.66 B6, multivitamin supplements, and colorectal cancer risk in women. Am J Epidemiol.
(2006) 15:108–15. doi: 10.1093/aje/kwj016
10. Gasbarrini A, Laterza L. La microbiota revolution: nuove conoscenze sul ruolo del
microbiota intestinale e possibili scenari nell’uso dei probiotici In: F Marangoni, P 35. Park S-Y, Wilkens LR, Setiawan VW, Monroe KR, Haiman CA, Le Marchand L.
Restani, L Morelli, A Gasbarrini, L Laterza and D Careddu, editors. Review Alcohol intake and colorectal cancer risk in the multiethnic cohort study. Am J
sull’integrazione alimentare: evidenza dalla ricerca scientifica e nuove frontiere di sviluppo. Epidemiol. (2019) 1:67–76. doi: 10.1093/aje/kwy208
2nd ed. Milano: Edra (2019). 44–8.
36. Botteri E, Borroni E, Sloan EK, Bagnardi V, Bosetti C, Peveri G, et al. Smoking and
11. Butel MJ. Probiotics, gut microbiota and health. Med Mal Infect. (2014) 44:1–8. colorectal cancer risk, overall and by molecular subtypes: A meta-analysis. Am J
doi: 10.1016/j.medmal.2013.10.002 Gastroenterol. (2020) 12:1940–9. doi: 10.14309/ajg.0000000000000803
12. Choden T, Cohen NA. The gut microbiome and the immune system. Expl Med. 37. Bardhan K, Liu K. Epigenetics and colorectal cancer pathogenesis. Cancers. (2013)
(2022) 3:219–33. doi: 10.37349/emed.2022.00087 5:676–713. doi: 10.3390/cancers5020676
13. Wu X, Tian J, Wang S. Insight into non-pathogenic Th17 cells in autoimmune 38. Wang T, Cai G, Qiu Y, Fei N, Zhang M, Pang X, et al. Structural segregation of gut
diseases. Front Immunol. (2018) 9:1112. doi: 10.3389/fimmu.2018.01112 microbiota between colorectal cancer patients and healthy volunteers. ISME J. (2012)
6:320–9. doi: 10.1038/ismej.2011.109
14. Ohkubo T, Tsuda M, Suzuki S, El Borai N, Yamamura M. Peripheral blood
neutrophils of germ-free rats modified by in vivo granulocyte-colony-stimulating factor 39. Wong SH, Zhao L, Zhang X, Nakatsu G, Han J, Xu W, et al. Gavage of fecal samples
and exposure to natural environment. Scand J Immunol. (1999) 49:73–7. doi: 10.1046/j. from patients with colorectal cancer promotes intestinal carcinogenesis in germfree and
1365-3083.1999.00456.x conventional mice. Gastroenterol. (2017) 153:1621–33. doi: 10.1053/j.gastro.2017.08.022
15. Qian LJ, Kang SM, Xie JL, Huang L, Wen Q, Fan YY, et al. Early-life gut microbial 40. Hou H, Chen D, Zhang K, Zhang W, Liu T, Wang S, et al. Gut microbiota derived
colonization shapes Th1/Th2 balance in asthma model in BALB/c mice. BMC Microbiol. short-fatty acids and colorectal cancer: ready for clinical transition. Cancer Lett. (2022)
(2017) 17:135. doi: 10.1186/s12866-017-1044-0 526:225–35. doi: 10.1016/j.canlet.2021.11.027
16. Li X, Zhang S, Guo G, Han J, Yu J. Gut microbiome in modulating immune 41. Torres-Maravilla E, Boucard AS, Mohseni AH, Taghinezhad SS, Cortes-Perez NG,
checkpoint inhibitors. Lancet. (2022) 82:104163. doi: 10.1016/j.ebiom.2022.104163 Bermudez-Humaran LG. Role of gut microbiota and probiotics in Colorectal Cancer:
Onset and progression. Microorganisms. (2021) 9:1021. doi: 10.3390/
17. Ye X, Li H, Anjum K, Zhong X, Miao S, Zheng G, et al. Dual role of indoles derived
microorganisms9051021
from intestinal microbiota on human health. Front Immunol. (2022) 13:903526. doi:
10.3389/fimmu.2022.903526 42. Deol PK, Khare P, Bishnoi M, Kondepudi KK, Kaur IP. Coadministration of ginger
extract–Lactobacillus acidophilus (cobiotic) reduces gut inflammation and oxidative
18. Morrison DJ, Preston T. Formation of short chain fatty acids by the gut microbiota
stress via downregulation of COX-2, i-NOS, and c-Myc. Phytother Res. (2018) 32:1950–6.
and their impact on human metabolism. Gut Microbes. (2016) 7:189–200. doi:
doi: 10.1002/ptr.6121
10.1080/19490976.2015.1134082

43. Ferreira-Halder CV, Faria AVS, Andrade SS. Action and function of
19. Salvi PS, Cowles RA. Butyrate and the intestinal epithelium: modulation of
Faecalibacterium prausnitzii in health and disease. Best Prat Res Cl Ga. (2017) 31:643–8.
proliferation and inflammation in homeostasis and disease. Cells. (2021) 10:1775. doi:
doi: 10.1016/j.bpg.2017.09.011
10.3390/cells10071775
44. Saito S, Kakizaki N, Okuno A, Maekawa T, Tsuji NM. Lactococcus lactis subsp.
20. Godlewska U, Burlanda E, Wypych TP. Bile acids in immunity: bidirectional cremoris C60 restores T cell population in small intestinal lamina propria in aged
mediators between the host and the microbiota. Front Immunol. (2022) 13:949033. doi: Interleukin-18 deficient mice. Nutrients. (2020) 12:3287. doi: 10.3390/nu12113287
10.3389/fimmu.2022.949033
45. Iwama T, Fujiva M, Konishi H, Tanaka H, Murakamu Y, Kuonogi T, et al. Bacteria-
21. Alexeev EE, Lanis JM, Kao DJ, Campbell EL, Kelly CJ, Battista KD, et al. derived ferrichrome inhibits tumor progression in sporadic colorectal neoplasms and
Microbiota-derived indole metabolites promote human and murine intestinal colitis-associated cancer. Cancer Cell Int. (2021) 21:21. doi: 10.1186/s12935-020-01723-9
homeostasis through regulation of Interleukin-10 receptor. Am J Pathol. (2018)
188:1183–94. doi: 10.1016/j.ajpath.2018.01.011 46. Shang F, Jiang X, Wang H, Chen S, Wang X, Liu Y, et al. The inhibitory effects of
probiotics on colon cancer cells: in vitro and in vivo studies. J Gastrointest Oncol. (2020)
22. Ehrlich AM, Pacheco AR, Henrick BM, Taft D, Xu G, Huda MN, et al. Indole-3- 11:1224–32. doi: 10.21037/jgo-20-573
lactic acid associated with Bifidobacterium-dominated microbiota significantly
decreases inflammation in intestinal epithelial cells. BMC Microbiol. (2020) 20:357. doi: 47. Dong Y, Zhu J, Zhang M, Ge S, Zhao L. Probiotic Lactobacillus salivarius Ren
10.1186/s12866-020-02023-y prevent dimethylhydrazine-induced colorectal cancer through protein kinase B
inhibition. Appl Microbiol Biotechnol. (2020) 104:7377–89. doi: 10.1007/
23. Ménard O, Butel MJ, Gaboriau-Routhiau V, Waligora-Dupriet AJ. Gnotobiotic s00253-020-10775-w
mouse immune response induced by Bifidobacterium sp. strains isolated from infants.
Appl Environ Microbiol. (2008) 74:660–6. doi: 10.1128/AEM.01261-07 48. Park IJ, Lee JH, Kye BH, Oh HK, Cho YB, Kim YT, et al. Effects of PrObiotics on
the symptoms and Surgical ouTComes after Anterior REsection of colon cancer
24. Ponz de Leon M, Rossi G, di Gregorio C, De Gaetani C, Rossi F, Ponti G, et al. (POSTCARE): A randomized, double-blind, placebo-controlled trial. J Clin Med. (2020)
Epidemiology of colorectal cancer: the 21-year of expirience of a specialised registry. 9:2181. doi: 10.3390/jcm9072181
Intern Emerg Med. (2007) 2:269–79. doi: 10.1007/s11739-007-0077-z
49. Yoon BJ, Oh HK, Lee J, Cho JR, Kim MJ, Kim DW, et al. Effects of probiotics on
25. Dionigi R. Chirurgia - Basi teoriche e Chirurgia generale - Chirurgia specialistica. bowel function restoration following ileostomy closure in rectal cancer patients: a
Milano: Masson (2019). randomized controlled trial. Color Dis. (2021) 23:901–10. doi: 10.1111/codi.15463
26. Rawla P, Sunkara T, Barsouk A. Epidemiology of colorectal cancer: incidence, 50. Sarkar A, Harty S, Lehto SM, Moeller AH, Dinan TG, Dunbar RIM, et al. The
mortality, survival, and risk factors. Gastroenterol. (2019) 14:89–103. doi: 10.5114/ microbiome in psychology and cognitive neuroscience. Trends Cogn Sci. (2018)
pg.2018.81072 22:611–36. doi: 10.1016/j.tics.2018.04.006
27. Grancher A, Michel P, Di Fiore F, Sefrioui D. Aspirin and colorectal cancer. Bull 51. Kelly JR, Borre Y, Obrien C, Patterson E, El Aidy S, Deane J, et al. Transferring the
Cancer. (2018) 105:171–80. doi: 10.1016/j.bulcan.2017.09.013 blues: depression-associated gut microbiota induces neurobehavioural changes in the
rat. J Psychiatr Res. (2016) 82:109–18. doi: 10.1016/j.jpsychires.2016.07.019
28. Sánchez-Alcoholado SC, Ordóñez R, Otero A, Plaza-Andrade I, Laborda-Illanes
A, Medina JA, et al. Gut microbiota-mediated inflammation and gut permeability in 52. Accettulli A, Corbo MR, Sinigaglia M, Speranza B, Campaniello D, Racioppo A,
patients with obesity and colorectal cancer. Int J Mol. (2020) 21:6782. doi: 10.3390/ et al. Psycho-Microbiology, a new frontier for probiotics: An exploratory overview.
ijms21186782 Microorganisms. (2022) 10:2141. doi: 10.3390/microorganisms10112141
29. Pan SY, DesMeules M. Energy intake, physical activity, energy balance, and cancer: 53. Wieërs G, Belkhir L, Enaud R, Leclercq S, De Foy JMP, Dequenne I, et al. How
epidemiologic evidence. Methods Mol Biol. (2009) 472:191–215. doi: probiotics affect the microbiota. Front Cell Infect Microbiol. (2020) 9:454. doi: 10.3389/
10.1007/978-1-60327-492-0_8 fcimb.2019.00454
30. Balhareth A, Aldossary MY, McNamara D. Impact of physical activity and diet on 54. Grimaldi R, Gibson G, Vulevic J, Giallourou N, Castro-Mejía J, Hansen L, et al. A
colorectal cancer survivors’ quality of life: a systematic review. World J Surg Oncol. (2019) prebiotic intervention study in children with autism spectrum disorders (ASDs).
17:17–153. doi: 10.1186/s12957-019-1697-2 Microbiome. (2018) 6:1–13. doi: 10.1186/s40168-018-0523-3
31. Frezza EE, Wachtel MS, Chiriva-Internati M. Influence of obesity on the risk of 55. Fattorusso A, Di Genova L, Dell’Isola GB, Mencaroni E, Esposito S. Autism spectrum
developing colon cancer. Gut. (2006) 55:285–91. doi: 10.1136/gut.2005.073163 disorders and the gut microbiota. Nutrients. (2019) 11:521. doi: 10.3390/nu11030521
32. Minkyeong K, Kyong P. Dietary Fat Intake and Risk of Colorectal Cancer: A 56. Grossi E, Melli S, Dunca D, Terruzzi V. Unexpected improvement in core autism
Systematic Review and Meta-Analysis of Prospective Studies. Nutrients. (2018) 10:1963. spectrum disorder symptoms after long-term treatment with probiotics. SAGE Open
doi: 10.3390/nu10121963 Med Case Rep. (2016) 4:1–5. doi: 10.1177/2050313X16666231

Frontiers in Nutrition 10 frontiersin.org


Campaniello et al. 10.3389/fnut.2023.1209238

57. Shaaban SY, El Gendy YG, Mehanna NS, El-Senousy WM, El-Feki HSA, Saad K, 78. Barichella M, Pacchetti C, Bolliri C, Cassani E, Iorio L, Pusani C, et al. Probiotics
et al. The role of probiotics in children with autism spectrum disorder: A prospective, and prebiotic fiber for constipation associated with Parkinson disease: An RCT.
open-label study. Nutr Neurosci. (2018) 21:676–81. doi: 10.1080/1028415X.2017.1347746 Neurology. (2016) 87:1274–80. doi: 10.1212/WNL.0000000000003127
58. Sanctuary MR, Kain JN, Chen SY, Kalanetra K, Lemay DG, Rose DR, et al. Pilot 79. Mendez-Figueroa V, Biscaia JM, Mohedano RB, Blanco-Fernandez A, Bailen M,
study of probiotic/colostrum supplementation on gut function in children with autism Bressa C, et al. Can gut microbiota and lifestyle help us in the handling of anorexia
and gastrointestinal symptoms. PLoS One. (2019) 14:e0210064. doi: 10.1371/journal. nervosa patients? Microorganisms. (2019) 7:58. doi: 10.3390/microorganisms7020058
pone.0210064
80. Achamrah N, Déchelotte P, Coëffier M. New therapeutic approaches to target gut-
59. Duque ALRF, Demarqui FM, Santoni MM, Zanelli CF, Adorno MAT, Dragan M, brain axis dysfunction during anorexia nervosa. Clin Nutr Exp. (2019) 28:33–41. doi:
et al. Effect of probiotic, prebiotic, and synbiotic on the gut microbiota of autistic 10.1016/j.yclnex.2019.01.006
children using an in vitro gut microbiome model. Food Res Int. (2021) 149:110657. doi:
81. Lew LC, Hor YY, Yusoff NAA, Choi SB, Yusoff MSB, Roslan NS, et al. Probiotic
10.1016/j.foodres.2021.110657
Lactobacillus plantarum P8 alleviated stress and anxiety while enhancing memory and
60. Liu YW, Liong MT, Chung YE, Huang HY, Peng WS, Cheng YF, et al. Effects of cognition in stressed adults: a randomised, double-blind, placebo-controlled study. Clin
Lactobacillus plantarum PS128 on children with autism spectrum disorder in Taiwan: a Nutr. (2018) 38:2053–64. doi: 10.1016/j.clnu.2018.09.010
randomized, double-blind, placebo-controlled trial. Nutrients. (2019) 11:820. doi:
82. Yano JM, Yu K, Donaldson GP, Shastri GG, Ann P, Ma L, et al. Indigenous bacteria
10.3390/nu11040820
from the gut microbiota regulate host serotonin biosynthesis. Cells. (2015) 161:264–76.
61. Kong XJ, Liu J, Li J, Kwong K, Koh M, Sukijthamapan P, et al. Probiotics and doi: 10.1016/j.cell.2015.02.047
oxytocin nasal spray as neuro-social-behavioral interventions for patients with autism
83. Angelakis E. Weight gain by gut microbiota manipulation in productive animals.
spectrum disorders: a pilot randomized controlled trial protocol. Pilot Feasibility Stud.
Microb Pathog. (2017) 106:162–70. doi: 10.1016/j.micpath.2016.11.002
(2020) 6:20. doi: 10.1186/s40814-020-0557-8
84. Marin IA, Goertz JE, Ren T, Rich SS, Onengut-Gumuscu S, Farber E, et al.
62. Pärtty A, Kalliomäki M, Wacklin P, Salminen S, Isolauri E. A possible link between
Microbiota alteration is associated with the development of stress-induced despair
early probiotic intervention and the risk of neuropsychiatric disorders later in childhood:
behavior. Sci Rep. (2017) 7:43859. doi: 10.1038/srep43859
A randomized trial. Pediatr Res. (2015) 77:823–8. doi: 10.1038/pr.2015.51

85. Cani PD, de Vos WM. Next-generation beneficial microbes: the case of
63. Cheng R, Xu W, Wang J, Tang Z, Zhang M. The outer membrane protein
Akkermansia muciniphila. Front Microbiol. (2017) 8:1765. doi: 10.3389/fmicb.2017.01765
Amuc_1100 of Akkermansia muciniphila alleviates the depression-like behavior of
depressed mice induced by chronic stress. Biochem Biophys Res Commun. (2021) 86. Rezaeiasl Z, Salami M, Sepehri G. The effects of probiotic Lactobacillus and
566:170–6. doi: 10.1016/j.bbrc.2021.06.018 Bifidobacterium strains on memory and learning behavior, long-term potentiation
(LTP), and some biochemical parameters in β-amyloid-induced rat’s model of
64. Abildgaard A, Elfving B, Hokland M, Wegener G, Lund S. Probiotic treatment
Alzheimer’s Disease. Prev Nutr Food Sci. (2019) 24:265–73. doi: 10.3746/
reduces depressive-like behaviour in rats independently of diet.
pnf.2019.24.3.265
Psychoneuroendocrinology. (2017) 79:40–8. doi: 10.1016/j.psyneuen.2017.02.014
87. Tamtaji OR, Heidari-Soureshjani R, Mirhosseini N, Kouchaki E, Bahmani F,
65. Reis DJ, Ilardi SS, Punt SEW. The anxiolytic effect of probiotics: a systematic review
Aghadavod E, et al. Probiotic and selenium co-supplementation, and the effects on
and meta-analysis of the clinical and preclinical literature. PLoS One. (2018)
clinical, metabolic and genetic status in Alzheimer’s disease: a randomized, double-
13:e0199041. doi: 10.1371/journal.pone.0199041
blind, controlled trial. Clin Nutr. (2019) 38:2569–75. doi: 10.1016/j.clnu.2018.11.034
66. Wallace CJK, Milev R. The effects of probiotics on depressive symptoms in
humans: a systematic review. Ann General Psychiatry. (2017) 16:14. doi: 10.1186/ 88. Dickerson FB, Stallings C, Origoni A, Katsafanas E, Savage CLG, Schweinfurth
s12991-017-0138-2 LAB, et al. Effect of probiotic supplementation on schizophrenia symptoms and
association with gastrointestinal functioning: a randomized, placebo-controlled trial.
67. Ghodarz Akkasheh G, Kashani-Poor Z, Tajabadi-Ebrahimi M, Jafari P, Akbari H, Prim Care Companion CNS Disord. (2014) 16:13m01579. doi: 10.4088/PCC.13m01579
Taghizadeh M, et al. Clinical and metabolic response to probiotic administration in
patients with major depressive disorder: a randomized, double-blind, placebo-controlled 89. Tankou SK, Regev K, Healy BC, Tjon E, Laghi L, Cox LM, et al. A probiotic
trial. Nutrition. (2016) 32:315–20. doi: 10.1016/j.nut.2015.09.003 modulates the microbiome and immunity in multiple sclerosis. Ann Neurol. (2018)
83:1147–61. doi: 10.1002/ana.25244
68. Liu RT, Walsh RFL, Sheehan AE. Prebiotics and probiotics for depression and
anxiety: a systematic review and meta-analysis of controlled clinical trials. Neurosci 90. Altieri C, Speranza B, Corbo MR, Sinigaglia M, Bevilacqua A. Gut-Microbiota,
Biobehav Rev. (2019) 102:13–23. doi: 10.1016/j.neubiorev.2019.03.023 and multiple sclerosis: Background, evidence, and perspectives. Nutrients. (2023) 15:942.
doi: 10.3390/nu15040942
69. Kazemi A, Noorbala AA, Azam K, Eskandari MH, Djafarian K. Effect of probiotic
and prebiotic vs placebo on psychological outcomes in patients with major depressive 91. Eales J, Gibson P, Whorwell P, Kellow J, Yellowlees A, Perry RHJ, et al. Systematic
disorder: A randomized clinical trial. Clin Nutr. (2019) 38:522–8. doi: 10.1016/j. review and meta-analysis: the effects of fermented milk with Bifidobacterium lactis
clnu.2018.04.010 CNCM I-2494 and lactic acid bacteria on gastrointestinal discomfort in the general adult
population. Therapeutic Adv Gastroenterol. (2017) 10:74–88. doi:

70. Kim CS, Cha L, Sim M, Jung S, Chun WY, Baik HW, et al. Probiotic 10.1177/1756283X16670075
supplementation improves cognitive function and mood with changes in gut microbiota
in community-dwelling older adults: A randomized, double-blind, placebo-controlled, 92. Lopetuso LR, Quagliarello A, Schiavoni M, Petito V, Russo A, Reddel S, et al.
multicenter trial. J Gerontol A Biol Sci Med Sci. (2021) 76:32–40. doi: 10.1093/gerona/ Towards a disease-associated common trait of gut microbiota dysbiosis: the pivotal role
glaa090 of Akkermansia muciniphila. Dig Liver Dis. (2020) 52:1002–10. doi: 10.1016/j.
dld.2020.05.020
71. Ma T, Jin H, Kwol LY, Sun Z, Liong MT, Zhang H. Probiotic consumption relieved
human stress and anxiety symptoms possibly via modulating the neuroactive potential 93. Ottman N, Geerlings SY, Aalvink S, de Vos WM, Belzer C. Action and function of
of the gut microbiota. Neurobiol Stress. (2021) 14:100294. doi: 10.1016/j. Akkermansia muciniphila in microbiome ecology, health and disease. Best Pract Res Clin
ynstr.2021.100294 Gastroenterol. (2017) 31:637–42. doi: 10.1016/j.bpg.2017.10.001
72. Romo-Araiza A, Gutierrez-Salmean G, Galvan EJ, Hernandez-Frausto M, Herrera- 94. Gallo A, Passaro G, Gasbarrini A, Landolfi R, Montalto M. Modulation of
Lopez G, Romo-Parra H, et al. Probiotics and prebiotics as a therapeutic strategy to microbiota as treatment for intestinal inflammatory disorders: an uptodate. World J
improve memory in a model of middle-aged rats. Front Aging Neurosci. (2018) 10:416. Gastroenterol. (2016) 22:7186–202. doi: 10.3748/wjg.v22.i32.7186
doi: 10.3389/fnagi.2018.00416
95. Azad MAK, Sarker M, Li T, Yin J. Probiotic species in the modulation of gut
73. Kobayashi Y, Kuhara T, Oki M, Xiao JZ. Effects of Bifidobacterium breve a1 on the microbiota: an overview. Biomed Res Int. (2018) 2018:9478630. doi:
cognitive function of older adults with memory complaints: a randomised, double-blind, 10.1155/2018/9478630
placebo-controlled trial. Benef Microbes. (2019) 10:511–20. doi: 10.3920/BM2018.0170
96. Dermyshi E, Wang Y, Yan C, Hong W, Qiu G, Gong X, et al. The “Golden Age” of
74. Ni Y, Yang X, Zheng L, Wang Z, Wu L, Jiang J, et al. Lactobacillus and probiotics: A systematic review and Meta-Analysis of randomized and observational
Bifidobacterium Improves physiological function and cognitive ability in aged mice by studies in preterm infants. Neonatology. (2017) 112:9–23. doi: 10.1159/000454668
the regulation of gut microbiota. Mol Nutr Food Res. (2019) 63:e1900603. doi: 10.1002/
97. Mezzasalma V, Manfrini E, Ferri E, Sandionigi A, La Ferla B, Schiano I, et al. A
mnfr.201900603
randomized, double-blind, placebo-controlled trial: The efficacy of multispecies
75. Liu J, Sun J, Wang F, Yu X, Ling Z, Li H, et al. Neuroprotective effects of Clostridium probiotic supplementation in alleviating symptoms of irritable bowel syndrome
butyricum against vascular dementia in mice via metabolic butyrate. Biomed Res Int. associated with constipation. Biomed Res Int. (2016) 2016:4740907. doi:
(2015) 2015:412946. doi: 10.1155/2015/412946 10.1155/2016/4740907
76. Tamtaji OR, Taghizadeh M, Daneshvar Kakhaki R, Kouchaki E, Bahmani F, 98. Majeed M, Nagabhushanam K, Natarajan S, Sivakumar A, Ali F, Pande A, et al.
Borzabadi S, et al. Clinical and metabolic response to probiotic administration in people Bacillus coagulans MTCC 5856 supplementation in the management of diarrhea
with Parkinson’s disease: a randomized, double-blind, placebo-controlled trial. Clin predominant irritable bowel syndrome: A double blind randomized placebo-controlled
Nutr. (2019) 38:1031–5. doi: 10.1016/j.clnu.2018.05.018 pilot clinical study. Nutr J. (2016) 15:21. doi: 10.1186/s12937-016-0140-6
77. Magistrelli L, Amoruso A, Mogna L, Graziano T, Cantello R, Pane M, et al. 99. Hemarajata P, Versalovic J. Effects of probiotics on gut microbiota: mechanisms of
Probiotics may have beneficial effects in Parkinson’s Disease: In vitro evidence. Front intestinal immunomodulation and neuromodulation. Therap Adv Gastroenterol. (2013)
Immunol. (2019) 10:969. doi: 10.3389/fimmu.2019.00969 6:39–51. doi: 10.1177/1756283X12459294

Frontiers in Nutrition 11 frontiersin.org


Campaniello et al. 10.3389/fnut.2023.1209238

100. Lyra A, Hillila M, Huttunen T, Mannikko S, Taalikka M, Tennila J, et al. Irritable 121. Kassaian N, Feizi A, Aminorroaya A, Amini M. Probiotic and synbiotic
bowel syndrome symptom severity improves equally with probiotic and placebo. World supplementation could improve metabolic syndrome in prediabetic adults: a randomized
J Gastroenterol. (2016) 22:10631–42. doi: 10.3748/wjg.v22.i48.10631 controlled trial. Diabetes Metab Syndr. (2019) 13:2991–6. doi: 10.1016/j.dsx.2018.07.016
101. Sisson G, Ayis S, Sherwood RA, Bjarnason I. Randomised clinical trial: A liquid 122. Porwal K, Pal S, Kulkarni C, Singh P, Sharma S, Singh P. A prebiotic, short-chain
multi-strain probiotic vs. placebo in the irritable bowel syndrome–A 12 week double- fructo-oligosaccharides promotes peak bone mass and maintains bone mass in
blind study. Aliment Pharmacol Ther. (2014) 40:51–62. doi: 10.1111/apt.12787 ovariectomized rats by an osteogenic mechanism. Biomed Pharmacother. (2020)
129:1–9. doi: 10.1016/j.biopha.2020.110448
102. Bervoets L, Van Hoorenbeeck K, Kortleven I, Van Noten C, Hens N, Vael C, et al.
Differences in gut microbiota composition between obese and lean children: a cross- 123. Hernandez CJ, Guss JD, Luna M, Goldring SR. Links between the microbiome
sectional study. Gut Pathog. (2013) 5:10. doi: 10.1186/1757-4749-5-10 and bone. J Bone Miner Res. (2016) 31:1638–46. doi: 10.1002/jbmr.2887
103. Kadooka Y, Sato M, Ogawa A, Miyoshi M, Uenishi H, Ogawa H, et al. Effect of 124. So JS, Song MK, Kwon HK, Lee CG, Chae CS, Sahoo A, et al. Lactobacillus casei
Lactobacillus gasseri SBT2055 in fermented milk on abdominal adiposity in adults in a enhances type II collagen/glucosamine-mediated suppression of inflammatory
randomised controlled trial. Br J Nutr. (2013) 110:1696–703. doi: 10.1017/ responses in experimental osteoarthritis. Life Sci. (2011) 88:358–66. doi: 10.1016/j.
S0007114513001037 lfs.2010.12.013
104. Omar JM, Chan YM, Jones ML, Prakash S, Jones PJH. Lactobacillus fermentum 125. Steves CJ, Bird S, Williams FM, Spector TD. The microbiome and musculoskeletal
and Lactobacillus amylovorus as probiotics alter body adiposity and gut microflora in conditions of aging: a review of evidence for impact and potential therapeutics. J Bone
healthy persons. J Funct Foods. (2013) 5:116–23. doi: 10.1016/j.jff.2012.09.001 Miner Res. (2016) 31:261–9. doi: 10.1002/jbmr.2765
105. Safavi M, Farajian S, Kelishadi R, Mirlohi M, Hashemipour M. The effects of 126. Paul AK, Paul A, Jahan R, Jannat K, Bondhon TA, Hasan A, et al. Probiotics and
synbiotic supplementation on some cardio-metabolic risk factors in overweight and amelioration of rheumatoid arthritis: significant roles of Lactobacillus casei and Lactobacillus
obese children: a randomized triple-masked controlled trial. Int J Food Sci Nutr. (2013) acidophilus. Microorganisms. (2021) 9:1070. doi: 10.3390/microorganisms9051070
64:687–93. doi: 10.3109/09637486.2013.775224
127. Gohel MK, Prajapati JB, Mudgal SV, Pandya HV, Singh US, Trivedi SS, et al. Effect
106. Asemi Z, Samimi M, Tabasi Z, Talebian P, Azarbad Z, Hydarzadeh Z, et al. Effect of probiotic dietary intervention on calcium and haematological parameters in
of daily consumption of probiotic yoghurt on lipid profiles in pregnant women: a geriatrics. J Clin Diagn Res. (2016) 10:5–9. doi: 10.7860/JCDR/2016/18877.7627
randomized controlled clinical trial. J Matern Fetal Neonatal Med. (2012) 25:1552–6.
128. Lei M, Hua LM, Wang DW. The effect of probiotic treatment on elderly patients
doi: 10.3109/14767058.2011.640372
with distal radius fracture: a prospective double-blind, placebo-controlled randomised
107. Jiang J, Wu C, Zhang C, Zhao J, Yu L, Zhang H, et al. Effects of probiotic clinical trial. Benef Microbes. (2016) 7:631–7. doi: 10.3920/BM2016.0067
supplementation on cardiovascular risk factors in hypercholesterolemia: A systematic
129. Han SK, Shin YJ, Lee DY, Kim KM, Yang SJ, Kim DS, et al. Lactobacillus
review and meta-analysis of randomized clinical trial. J Funct Foods. (2020) 74:104–77.
rhamnosus HDB1258 modulates gut microbiota-mediated immune response in mice
doi: 10.1016/j.jff.2020.104177
with or without lipopolysaccharide-induced systemic inflammation. BMC Microbiol.
108. Corb Aron RAC, Abid A, Vesa CM, Nechifor AC, Behl T, Ghitea TC, et al. (2021) 21:146. doi: 10.1186/s12866-021-02192-4
Recognizing the benefits of pre−/probiotics in metabolic syndrome and type 2 diabetes
130. National Institute of Health. Office of Dietary Supplements Probiotics. Fact sheets
mellitus considering the influence of Akkermansia muciniphila as a key gut bacterium.
for health professionals. (2022). Available at: https://ods.od.nih.gov/factsheets/Probiotics-
Microorganisms. (2021) 9:618. doi: 10.3390/microorganisms9030618
HealthProfessional/ (Accessed June 14, 2023).
109. Blaak EE, Canfora EE, Theis S, Frost G, Groen AK, Mitheus G, et al. Short chain
131. Hathi Z, Mettu S, Priya A, Athukoralalage S, Lam TN, Choudhury NR, et al.
fatty acids in human gut and metabolic health. Benef Microbes. (2020) 11:411–55. doi:
Methodological advances and challenges in probiotic bacteria production: Ongoing
10.3920/BM2020.0057
strategies and future perspectives. Biochem Eng J. (2021) 176:108199. doi: 10.1016/j.
110. Portincasa P, Bonfrate L, Vacca M, De Angelis M, Farella I, Lanza E, et al. Gut bej.2021.108199
microbiota and short chain fatty acids: implications in glucose homeostasis. Int J Mol
132. International Association for Porbiotics and Prebiotics. Deciphering a probiotic
Sci. (2022) 23:1105. doi: 10.3390/ijms23031105
label. (2022). Available at: http://isappscience.org/wp-content/uploads/2019/04/EU_
111. Alsharairi NA. The role of short-chain fatty acids in mediating very low-calorie Probiotic_labeling_rev1029.pdf (Accessed June 14, 2023).
ketogenic diet-infant gut microbiota relationships and its therapeutic potential in 133. Islam SUI. Clinical uses of probiotics. Medicine. (2016) 95:5. doi: 10.1097/
obesity. Nutrients. (2021) 13:3702. doi: 10.3390/nu13113702 MD.0000000000002658
112. Gurung M, Li Z, You H, Rodrigues R, Jump DB, Morgun A, et al. Role of gut 134. Miller LE, Lehtoranta L, Lehtinen MJ. Short-term probiotic supplementation
microbiota in type 2 diabetes pathophysiology. EBioMedicine. (2020):51102590. doi: enhances cellular immune function in healthy elderly: systematic review and meta-
10.1016/j.ebiom.2019.11.051 analysis of controlled studies. Nutr Res. (2016) 64:1–8. doi: 10.1016/j.nutres.2018.12.011
113. Wang S, Xiao Y, Tian F, Zhao J, Zhang H, Chen W. Rational use of prebiotics for 135. Forth E, Buehner B, Storer A, Sgarbossa C, Milev R, Meyyappan AC. Systematic
gut microbiota alterations: specific bacterial phylotypes and related mechanisms. J Funct review of probiotics as adjuvant treatment for psychiatric disorders. Front Behav
Foods. (2020) 66:1–13. doi: 10.1016/j.jff.2020.103838 Neurosci. (2023) 17:1111349. doi: 10.3389/fnbeh.2023.1111349
114. Mobini R, Tremaroli V, Ståhlman M, Karlsson F, Levin M, Ljungberg M, et al. 136. Amirami E, Milajerdi A, Mirzaeih JH, Mansournia MA, Hallajzadeh J, et al. The
Metabolic effects of Lactobacillus reuteri DSM 17938 in people with type 2 diabetes: a effects of probiotic supplementation on mental health, biomarkers of inflammation and
randomized controlled trial. Diabetes Obes Metab. (2017) 19:579–89. doi: 10.1111/ oxidative stress in patients with psychiatric disorders: A systematic review and meta-
dom.12861 analysis of randomized controlled trials. Complement Ther Med. (2023) 49:102361. doi:
10.1016/j.ctim.2020.102361
115. Toejing P, Khampithum N, Sirilun S, Chaiyasut C, Lailerd N. Influence of
Lactobacillus paracasei HII01 supplementation on glycemia and inflammatory 137. Baral KC, Bajracharya R, Lee SH, Han H-K. Advancements in the pharmaceutical
biomarkers in type 2 diabetes: A randomized clinical trial. Foods. (2021) 10:1455. doi: applications of probiotics: dosage forms and formulation technology. Int J Nanomedicine.
10.3390/foods10071455 (2021) 16:7535–56. doi: 10.2147/IJN.S337427
116. Wu T, Zhang Y, Li W, Zhao Y, Long H, Muhindo EM, et al. Lactobacillus 138. Cunningham M, Vinderola G, Charalampopoulos D, Leeber S, Sanders ME,
rhamnosus LRa05 Ameliorate hyperglycemia trough a regulating glucagon-mediated Grimaldi R. Applying probiotics and prebiotics in new delivery formats – is the clinical
signaling pathway and gut microbiota in type 2 diabetic mice. J Agric Food Chem. (2021) evidence transferable? Trends Food Sci Technol. (2021) 11:495–506. doi: 10.1016/j.
69:8797–806. doi: 10.1021/acs.jafc.1c02925 tifs.2021.04.009
117. Manaer T, Yu L, Nabi XH, Dilidaxi D, Liu L, Sailike J. The beneficial effects of the 139. Santocchi E, Guiducci L, Prosperi M, Calderoni S, Gaggini M, Apicella F, et al.
composite of probiotics from camel milk on glucose and lipid metabolism, liver and Effects of probiotic supplementation on gastrointestinal, sensory and core symptoms in
renal function and gut microbiota in db/db mice. BMC Complement Med Ther. (2021) autism spectrum disorders: a randomized controlled trial. Front Psych. (2020) 11:550593.
21:127. doi: 10.1186/s12906-021-03303-4 doi: 10.3389/fpsyt.2020.550593
118. Razmpoosh E, Javadi A, Ejtahed HS, Mirmiran P, Javadi M, Yousefinejad A. The 140. Tomova A, Husarova V, Lakatosova S, Bakos J, Vlkova B, Babinska K, et al.
effect of probiotic supplementation on glycemic control and lipid profile in patients with Gastrointestinal microbiota in children with autism in Slovakia. Physiol Behav. (2015)
type 2 diabetes: a randomized placebo controlled trial. Diabetes Metab Syndr. (2019) 138:179–87. doi: 10.1016/j.physbeh.2014.10.033
13:175–82. doi: 10.1016/j.dsx.2018.08.008
141. Wang X, Yang J, Zhang H, Yu J, Yao Z. Oral probiotic administration during
119. Zheng J, Cheng G, Li Q, Jiao S, Feng C, Zhao X, et al. Chitin oligosaccharide pregnancy prevents autism-related behaviors in offspring induced by maternal immune
modulates gut microbiota and attenuates high-fat-diet-induced metabolic syndrome in activation via anti-inflammation in mice. Autism Res. (2019) 12:576–88. doi: 10.1002/
mice. Mar Drugs. (2018) 16:66. doi: 10.3390/md16020066 aur.2079
120. Barreto FM, Colado Simão AN, Morimoto HK, Lozovoy MAB, Dichi MDI, da 142. El-Ansary A, Bacha AB, Bjørklund G, Al-Orf N, Bhat RS, Moubayed N, et al.
Silva H, et al. Beneficial effects of Lactobacillus plantarum on glycemia and homocysteine Probiotic treatment reduces the autistic-like excitation/inhibition imbalance in juvenile
levels in postmenopausal women with metabolic syndrome. Nutrition. (2014) 30:939–42. hamsters induced by orally administered propionic acid and clindamycin. Metab Brain
doi: 10.1016/j.nut.2013.12.004 Dis. (2018) 331155–64. doi: 10.1007/s11011-018-0212-8

Frontiers in Nutrition 12 frontiersin.org

You might also like